Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Small ; 20(6): e2306387, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37771189

RESUMEN

4D printing recently emerges as an exciting evolution of conventional 3D printing, where a printed construct can quickly transform in response to a specific stimulus to switch between a temporary variable state and an original state. In this work, a photocrosslinkable polyethylene-glycol polyurethane ink is synthesized for light-assisted 4D printing of smart materials. The molecular weight distribution of the ink monomers is tunable by adjusting the copolymerization reaction time. Digital light processing (DLP) technique is used to program a differential swelling response in the printed constructs after humidity variation. Bioactive microparticles are embedded into the ink and the improvement of biocompatibility of the printed constructs is demonstrated for tissue engineering applications. Cell studies reveal above 90% viability in 1 week and ≈50% biodegradability after 4 weeks. Self-folding capillary scaffolds, dynamic grippers, and film actuators are made and activated in a humid environment. The approach offers a versatile platform for the fabrication of complex constructs. The ink can be used in tissue engineering and actuator applications, making the ink a promising avenue for future research.


Asunto(s)
Tinta , Andamios del Tejido , Poliuretanos , Ingeniería de Tejidos/métodos , Hidrogeles , Impresión Tridimensional
2.
J Mol Cell Cardiol ; 168: 3-12, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35390437

RESUMEN

Engineered heart muscle (EHM) can be implanted epicardially to remuscularize the failing heart. In case of a severely scarred ventricle, excision of scar followed by transmural heart wall replacement may be a more desirable application. Accordingly, we tested the hypothesis that allograft (rat) and xenograft (human) EHM can also be administered as transmural heart wall replacement in a heterotopic, volume-loaded heart transplantation model. We first established a novel rat model model to test surgical transmural left heart wall repair. Subsequently and in continuation of our previous allograft studies, we tested outcome after implantation of contractile engineered heart muscle (EHM) and non-contractile engineered connective tissue (ECT) as well as engineered mesenchymal tissue (EMT) allografts as transmural heart wall replacement. Finally, proof-of-concept for the application of human EHM was obtained in an athymic nude rat model. Only in case of EHM implantation, remuscularization of the surgically created transmural defect was observed with palpable graft vascularization. Taken together, feasibility of transmural heart repair using bioengineered myocardial grafts could be demonstrated in a novel rat model of heterotopic heart transplantation.


Asunto(s)
Trasplante de Corazón , Miocitos Cardíacos , Animales , Humanos , Miocardio , Miocitos Cardíacos/fisiología , Ratas , Ratas Desnudas , Ingeniería de Tejidos
3.
Circulation ; 142(11): 1059-1076, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32623905

RESUMEN

BACKGROUND: Noonan syndrome (NS) is a multisystemic developmental disorder characterized by common, clinically variable symptoms, such as typical facial dysmorphisms, short stature, developmental delay, intellectual disability as well as cardiac hypertrophy. The underlying mechanism is a gain-of-function of the RAS-mitogen-activated protein kinase signaling pathway. However, our understanding of the pathophysiological alterations and mechanisms, especially of the associated cardiomyopathy, remains limited and effective therapeutic options are lacking. METHODS: Here, we present a family with two siblings displaying an autosomal recessive form of NS with massive hypertrophic cardiomyopathy as clinically the most prevalent symptom caused by biallelic mutations within the leucine zipper-like transcription regulator 1 (LZTR1). We generated induced pluripotent stem cell-derived cardiomyocytes of the affected siblings and investigated the patient-specific cardiomyocytes on the molecular and functional level. RESULTS: Patients' induced pluripotent stem cell-derived cardiomyocytes recapitulated the hypertrophic phenotype and uncovered a so-far-not-described causal link between LZTR1 dysfunction, RAS-mitogen-activated protein kinase signaling hyperactivity, hypertrophic gene response and cellular hypertrophy. Calcium channel blockade and MEK inhibition could prevent some of the disease characteristics, providing a molecular underpinning for the clinical use of these drugs in patients with NS, but might not be a sustainable therapeutic option. In a proof-of-concept approach, we explored a clinically translatable intronic CRISPR (clustered regularly interspaced short palindromic repeats) repair and demonstrated a rescue of the hypertrophic phenotype. CONCLUSIONS: Our study revealed the human cardiac pathogenesis in patient-specific induced pluripotent stem cell-derived cardiomyocytes from NS patients carrying biallelic variants in LZTR1 and identified a unique disease-specific proteome signature. In addition, we identified the intronic CRISPR repair as a personalized and in our view clinically translatable therapeutic strategy to treat NS-associated hypertrophic cardiomyopathy.


Asunto(s)
Sistemas CRISPR-Cas , Cardiomiopatías , Terapia Genética , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Cardiovasculares , Mutación , Miocitos Cardíacos/metabolismo , Síndrome de Noonan , Factores de Transcripción , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/terapia , Humanos , Intrones , Síndrome de Noonan/genética , Síndrome de Noonan/metabolismo , Síndrome de Noonan/terapia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Annu Rev Pharmacol Toxicol ; 58: 65-82, 2018 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-29029591

RESUMEN

Enhancing the early detection of new therapies that are likely to carry a safety liability in the context of the intended patient population would provide a major advance in drug discovery. Microphysiological systems (MPS) technology offers an opportunity to support enhanced preclinical to clinical translation through the generation of higher-quality preclinical physiological data. In this review, we highlight this technological opportunity by focusing on key target organs associated with drug safety and metabolism. By focusing on MPS models that have been developed for these organs, alongside other relevant in vitro models, we review the current state of the art and the challenges that still need to be overcome to ensure application of this technology in enhancing drug discovery.


Asunto(s)
Descubrimiento de Drogas/métodos , Preparaciones Farmacéuticas/química , Animales , Evaluación Preclínica de Medicamentos/métodos , Humanos
5.
Handb Exp Pharmacol ; 265: 143-156, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33136187

RESUMEN

Classical drug development is compromised by considerable clinical failure of promising drug candidates after decades of costly preclinical work. Failure can be because of previously unrecognized safety concerns or more commonly lack of clinical efficacy. Classical drug discovery and safety pharmacology programs rely heavily on well-established in vitro and preclinical animal models. The availability of human pluripotent stem cells and the possibility to direct them into any somatic cell type suggest that a paradigm shift in drug development may be possible and timely, with the opportunity to test safety and efficacy of candidate drugs on the human target cells and tissue. However, there is considerable uncertainty as to whether human models would only qualify as replacement for well-established tools or add substantially more information to the preclinical data package, to facilitate translation of more promising drug candidates into clinical practice. This chapter provides an overview of tissue-engineered macro-scale heart muscle models for applications in drug discovery and safety pharmacology.


Asunto(s)
Preparaciones Farmacéuticas , Células Madre Pluripotentes , Animales , Evaluación Preclínica de Medicamentos , Humanos , Miocardio , Miocitos Cardíacos
6.
J Mol Cell Cardiol ; 134: 13-28, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31233754

RESUMEN

Cardiac fibrosis is a hallmark of heart failure for which there is no effective pharmacological therapy. By genetic modification and in vivo inhibitor approaches it was suggested that the Rho-associated kinases (ROCK1 and ROCK2) are involved in pro-fibrotic signalling in cardiac fibroblasts and that they may serve as targets for anti-fibrotic therapies. We demonstrate that simultaneous inhibition of ROCK1 and ROCK2 strongly interfered with tissue formation and their biomechanical properties in a model of engineered connective tissue (ECT), comprised of cardiac fibroblasts and collagen. These effects were observed with both rat and human ECT. Inhibitors of different chemistries, including the isoquinoline inhibitors Fasudil and H1152P as well as the pyrazol-phenyl inhibitor SR-3677, showed comparable effects. By combined treatment of ECT with TGF-ß and H1152P, we could identify ROCK as a mediator of TGF-ß-dependent tissue stiffening. Moreover, expression analyses suggested that lysyl oxidase (LOX) is a downstream target of the ROCK-actin-MRTF/SRF pathway and inhibition of this pathway by Latrunculin A and CCG-203971 showed similar anti-fibrotic effects in the ECT model as ROCK inhibitors. In line with the collagen crosslinking function of LOX, its inhibition by ß-aminopropionitrile resulted in reduced ECT stiffness, but let tissue compaction unaffected. Finally, we show that ROCK inhibition also reduced the compaction and stiffness of engineered heart muscle tissues. Our results indicate that pharmacological inhibition of ROCK has a strong anti-fibrotic potential which is in part due to a decrease in the expression of the collagen crosslinking enzyme lysyl oxidase.


Asunto(s)
Miocardio/metabolismo , Miofibroblastos/metabolismo , Quinasas Asociadas a rho/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Actinas/metabolismo , Aminopropionitrilo/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Fibrosis/metabolismo , Corazón/efectos de los fármacos , Humanos , Masculino , Miofibroblastos/efectos de los fármacos , Proteína-Lisina 6-Oxidasa/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
7.
Europace ; 21(9): 1410-1421, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31106349

RESUMEN

AIMS: Brugada syndrome (BrS) is associated with a pronounced risk to develop sudden cardiac death (SCD). Up to 21% of patients are related to mutations in SCN5A. Studies identified SCN10A as a contributor of BrS. However, the investigation of the human cellular phenotype of BrS in the presence of SCN10A mutations remains lacking. The objective of this study was to establish a cellular model of BrS in presence of SCN10A mutations using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). METHODS AND RESULTS: Dermal fibroblasts obtained from a BrS patient suffering from SCD harbouring the SCN10A double variants (c.3803G>A and c.3749G>A) and three independent healthy control subjects were reprogrammed to hiPSCs. Human-induced pluripotent stem cells were differentiated into cardiomyocytes (hiPSC-CMs).The hiPSC-CMs from the BrS patient showed a significantly reduced peak sodium channel current (INa) and a significantly reduced ATX II (sea anemone toxin, an enhancer of late INa) sensitive as well as A-887826 (a blocker of SCN10A channel) sensitive late sodium channel current (INa) when compared with the healthy control hiPSC-CMs, indicating loss-of-function of sodium channels. Consistent with reduced INa the action potential amplitude and upstroke velocity (Vmax) were significantly reduced, which may contribute to arrhythmogenesis of BrS. Moreover, Ajmaline effects on action potentials were stronger in BrS-hiPSC-CMs than in healthy control cells. This is in agreement with the higher susceptibility of patients to sodium channel blocking drugs in unmasking BrS. CONCLUSION: Patient-specific hiPSC-CMs are able to recapitulate single-cell phenotype features of BrS with SCN10A mutations and may provide novel opportunities to further elucidate the cellular disease mechanism.


Asunto(s)
Potenciales de Acción/fisiología , Síndrome de Brugada/genética , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8/genética , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Ajmalina/farmacología , Síndrome de Brugada/metabolismo , Cardiotónicos/farmacología , Estudios de Casos y Controles , Técnicas de Reprogramación Celular , Venenos de Cnidarios/farmacología , Muerte Súbita Cardíaca , Humanos , Células Madre Pluripotentes Inducidas , Mutación con Pérdida de Función , Masculino , Persona de Mediana Edad , Morfolinas/farmacología , Mutación , Miocitos Cardíacos/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Niacinamida/análogos & derivados , Niacinamida/farmacología , Técnicas de Placa-Clamp , Fenotipo , Taquicardia Ventricular , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología
8.
Circulation ; 135(19): 1832-1847, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28167635

RESUMEN

BACKGROUND: Advancing structural and functional maturation of stem cell-derived cardiomyocytes remains a key challenge for applications in disease modeling, drug screening, and heart repair. Here, we sought to advance cardiomyocyte maturation in engineered human myocardium (EHM) toward an adult phenotype under defined conditions. METHODS: We systematically investigated cell composition, matrix, and media conditions to generate EHM from embryonic and induced pluripotent stem cell-derived cardiomyocytes and fibroblasts with organotypic functionality under serum-free conditions. We used morphological, functional, and transcriptome analyses to benchmark maturation of EHM. RESULTS: EHM demonstrated important structural and functional properties of postnatal myocardium, including: (1) rod-shaped cardiomyocytes with M bands assembled as a functional syncytium; (2) systolic twitch forces at a similar level as observed in bona fide postnatal myocardium; (3) a positive force-frequency response; (4) inotropic responses to ß-adrenergic stimulation mediated via canonical ß1- and ß2-adrenoceptor signaling pathways; and (5) evidence for advanced molecular maturation by transcriptome profiling. EHM responded to chronic catecholamine toxicity with contractile dysfunction, cardiomyocyte hypertrophy, cardiomyocyte death, and N-terminal pro B-type natriuretic peptide release; all are classical hallmarks of heart failure. In addition, we demonstrate the scalability of EHM according to anticipated clinical demands for cardiac repair. CONCLUSIONS: We provide proof-of-concept for a universally applicable technology for the engineering of macroscale human myocardium for disease modeling and heart repair from embryonic and induced pluripotent stem cell-derived cardiomyocytes under defined, serum-free conditions.


Asunto(s)
Células Madre Embrionarias/trasplante , Insuficiencia Cardíaca/terapia , Células Madre Pluripotentes Inducidas/trasplante , Miocitos Cardíacos/trasplante , Ingeniería de Tejidos/métodos , Remodelación Ventricular/fisiología , Animales , Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Insuficiencia Cardíaca/patología , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Miocardio/citología , Miocardio/patología , Miocitos Cardíacos/fisiología , Impresión Tridimensional , Ratas , Ratas Desnudas
9.
Circ Res ; 128(1): 39-41, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33411630

Asunto(s)
Fibroblastos , Corazón
10.
Europace ; 20(FI1): f46-f56, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29566126

RESUMEN

Aims: Our aim is to investigate the arrhythmogenic mechanism in arrhythmogenic right ventricular cardiomyopathy (ARVC)-patients by using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Methods and results: Human-induced pluripotent stem cell-derived cardiomyocytes were generated from human skin fibroblasts of two healthy donors and an ARVC-patient with a desmoglein-2 (DSG2) mutation. Patch clamp, quantitative polymerase chain reaction, and calcium imaging techniques were employed for the study. The amplitude and maximal upstroke velocity (Vmax) of action potential (AP) in ARVC-cells were smaller than that in healthy donor cells, whereas the resting potential and AP duration (APD) was not changed. The reduced Vmax resulted from decreased peak sodium current. The reason for undetected changes in APD may be the counter-action of reduced transient outward, small conductance Ca2+-activated, adenosine triphosphate-sensitive, Na/Ca exchanger (INCX) currents, and enhanced rapidly delayed rectifier currents. Isoprenaline (Iso) reduced INCX and shortened APD in both donor and ARVC-hiPSC-CMs. However, the effects of Iso in ARVC-cells are significantly larger than that in donor cells. In addition, ARVC-hiPSC-CMs showed more frequently than donor cells arrhythmogenic events induced by adrenergic stimulation. Conclusion: Cardiomyocytes derived from the ARVC patient with a DSG2 mutation displayed multiple ion channel dysfunctions and abnormal cellular electrophysiology as well as enhanced sensitivity to adrenergic stimulation. These may underlie the arrhythmogenesis in ARVC patients.


Asunto(s)
Potenciales de Acción , Displasia Ventricular Derecha Arritmogénica/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Potenciales de Acción/efectos de los fármacos , Agonistas Adrenérgicos beta/farmacología , Displasia Ventricular Derecha Arritmogénica/genética , Displasia Ventricular Derecha Arritmogénica/patología , Displasia Ventricular Derecha Arritmogénica/fisiopatología , Señalización del Calcio , Estudios de Casos y Controles , Células Cultivadas , Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Desmogleína 2/genética , Desmogleína 2/metabolismo , Predisposición Genética a la Enfermedad , Frecuencia Cardíaca , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/patología , Isoproterenol/farmacología , Cinética , Masculino , Persona de Mediana Edad , Mutación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fenotipo , Intercambiador de Sodio-Calcio/metabolismo
11.
J Mol Cell Cardiol ; 113: 9-21, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28941705

RESUMEN

The ability to generate patient-specific induced pluripotent stem cells (iPSCs) provides a unique opportunity for modeling heart disease in vitro. In this study, we generated iPSCs from a patient with dilated cardiomyopathy (DCM) caused by a missense mutation S635A in RNA-binding motif protein 20 (RBM20) and investigated the functionality and cell biology of cardiomyocytes (CMs) derived from patient-specific iPSCs (RBM20-iPSCs). The RBM20-iPSC-CMs showed abnormal distribution of sarcomeric α-actinin and defective calcium handling compared to control-iPSC-CMs, suggesting disorganized myofilament structure and altered calcium machinery in CMs of the RBM20 patient. Engineered heart muscles (EHMs) from RBM20-iPSC-CMs showed that not only active force generation was impaired in RBM20-EHMs but also passive stress of the tissue was decreased, suggesting a higher visco-elasticity of RBM20-EHMs. Furthermore, we observed a reduced titin (TTN) N2B-isoform expression in RBM20-iPSC-CMs by demonstrating a reduction of exon skipping in the PEVK region of TTN and an inhibition of TTN isoform switch. In contrast, in control-iPSC-CMs both TTN isoforms N2B and N2BA were expressed, indicating that the TTN isoform switch occurs already during early cardiogenesis. Using next generation RNA sequencing, we mapped transcriptome and splicing target profiles of RBM20-iPSC-CMs and identified different cardiac gene networks in response to the analyzed RBM20 mutation in cardiac-specific processes. These findings shed the first light on molecular mechanisms of RBM20-dependent pathological cardiac remodeling leading to DCM. Our data demonstrate that iPSC-CMs coupled with EHMs provide a powerful tool for evaluating disease-relevant functional defects and for a deeper mechanistic understanding of alternative splicing-related cardiac diseases.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Adulto , Animales , Calcio/metabolismo , Células Cultivadas , Conectina/metabolismo , Femenino , Humanos , Ratones , Mutación , Fenotipo , Empalme del ARN/genética , Sarcómeros/metabolismo , Transcriptoma/genética
12.
J Physiol ; 595(12): 3685-3690, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28295371

RESUMEN

Myocardial remuscularization can be achieved by cardiomyocyte implantation. Electromechanical integration and long-term survival of cardiomyocyte grafts are essential for maximal therapeutic impact. Cardiomyocyte application with support material has been instrumental in enhancing cell retention. Co-administration of pro-survival factors and immunological matching are additional strategies for increased cell graft survival. Finally, larger cardiomyocyte grafts, although therapeutically desirable, will increase the risk for arrhythmias and, if pluripotent stem cells are used to derive cardiomyocytes, tumour formation. This review introduces major challenges pertaining to myocardial remuscularization (cardiomyocyte retention, arrhythmogenicity and tumourigenicity), discusses studies addressing these challenges, and suggests strategies to overcome remaining challenges for the translation of myocardial remuscularization.


Asunto(s)
Insuficiencia Cardíaca/terapia , Miocardio/citología , Miocitos Cardíacos/fisiología , Regeneración/fisiología , Animales , Arritmias Cardíacas/terapia , Supervivencia Celular/fisiología
13.
Circ Res ; 117(5): 401-12, 2015 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-26078285

RESUMEN

RATIONALE: Monitoring and controlling cardiac myocyte activity with optogenetic tools offer exciting possibilities for fundamental and translational cardiovascular research. Genetically encoded voltage indicators may be particularly attractive for minimal invasive and repeated assessments of cardiac excitation from the cellular to the whole heart level. OBJECTIVE: To test the hypothesis that cardiac myocyte-targeted voltage-sensitive fluorescence protein 2.3 (VSFP2.3) can be exploited as optogenetic tool for the monitoring of electric activity in isolated cardiac myocytes and the whole heart as well as function and maturity in induced pluripotent stem cell-derived cardiac myocytes. METHODS AND RESULTS: We first generated mice with cardiac myocyte-restricted expression of VSFP2.3 and demonstrated distinct localization of VSFP2.3 at the t-tubulus/junctional sarcoplasmic reticulum microdomain without any signs for associated pathologies (assessed by echocardiography, RNA-sequencing, and patch clamping). Optically recorded VSFP2.3 signals correlated well with membrane voltage measured simultaneously by patch clamping. The use of VSFP2.3 for human action potential recordings was confirmed by simulation of immature and mature action potentials in murine VSFP2.3 cardiac myocytes. Optical cardiograms could be monitored in whole hearts ex vivo and minimally invasively in vivo via fiber optics at physiological heart rate (10 Hz) and under pacing-induced arrhythmia. Finally, we reprogrammed tail-tip fibroblasts from transgenic mice and used the VSFP2.3 sensor for benchmarking functional and structural maturation in induced pluripotent stem cell-derived cardiac myocytes. CONCLUSIONS: We introduce a novel transgenic voltage-sensor model as a new method in cardiovascular research and provide proof of concept for its use in optogenetic sensing of physiological and pathological excitation in mature and immature cardiac myocytes in vitro and in vivo.


Asunto(s)
Potenciales de la Membrana/fisiología , Miocitos Cardíacos/fisiología , Optogenética/métodos , Animales , Humanos , Ratones , Ratones Transgénicos , Imagen de Colorante Sensible al Voltaje/métodos
14.
Mol Ther ; 29(10): 2894-2895, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34555312
15.
Curr Cardiol Rep ; 19(9): 78, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28752277

RESUMEN

PURPOSE OF REVIEW: This review provides an overview of the current state of tissue-engineered heart repair with a special focus on the anticipated modes of action of tissue-engineered therapy candidates and particular implications as to transplant immunology. RECENT FINDINGS: Myocardial tissue engineering technologies have made tremendous advances in recent years. Numerous different strategies are under investigation and have reached different stages on their way to clinical translation. Studies in animal models demonstrated that heart repair requires either remuscularization by delivery of bona fide cardiomyocytes or paracrine support for the activation of endogenous repair mechanisms. Tissue engineering approaches result in enhanced cardiomyocyte retention and sustained remuscularization, but may also be explored for targeted paracrine or mechanical support. Some of the more advanced tissue engineering approaches are already tested clinically; others are at late stages of pre-clinical development. Process optimization towards cGMP compatibility and clinical scalability of contractile engineered human myocardium is an essential step towards clinical translation. Long-term allograft retention can be achieved under immune suppression. HLA matching may be an option to enhance graft retention and reduce the need for comprehensive immune suppression. Tissue-engineered heart repair is entering the clinical stage of the translational pipeline. Like in any effective therapy, side effects must be anticipated and carefully controlled. Allograft implantation under immune suppression is the most likely clinical scenario. Strategies to overcome transplant rejection are evolving and may further boost the clinical acceptance of tissue-engineered heart repair.


Asunto(s)
Insuficiencia Cardíaca/terapia , Miocardio , Miocitos Cardíacos , Ingeniería de Tejidos/métodos , Animales , Humanos
18.
J Mol Cell Cardiol ; 88: 39-54, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26392029

RESUMEN

Cardiac remodeling, a hallmark of heart disease, is associated with intense auto- and paracrine signaling leading to cardiac fibrosis. We hypothesized that the specific mediator of Gq/11-dependent RhoA activation p63RhoGEF, which is expressed in cardiac fibroblasts, plays a role in the underlying processes. We could show that p63RhoGEF is up-regulated in mouse hearts subjected to transverse aortic constriction (TAC). In an engineered heart muscle model (EHM), p63RhoGEF expression in cardiac fibroblasts increased resting and twitch tensions, and the dominant negative p63ΔN decreased both. In an engineered connective tissue model (ECT), p63RhoGEF increased tissue stiffness and its knockdown as well as p63ΔN reduced stiffness. In 2D cultures of neonatal rat cardiac fibroblasts, p63RhoGEF regulated the angiotensin II (Ang II)-dependent RhoA activation, the activation of the serum response factor, and the expression and secretion of the connective tissue growth factor (CTGF). All these processes were inhibited by the knockdown of p63RhoGEF or by p63ΔN likely based on their negative influence on the actin cytoskeleton. Moreover, we show that p63RhoGEF also regulates CTGF in engineered tissues and correlates with it in the TAC model. Finally, confocal studies revealed a closely related localization of p63RhoGEF and CTGF in the trans-Golgi network.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/genética , Fibroblastos/metabolismo , Miocardio/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factor de Respuesta Sérica/genética , Proteína de Unión al GTP rhoA/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Angiotensina II/genética , Angiotensina II/metabolismo , Animales , Animales Recién Nacidos , Aorta/cirugía , Comunicación Autocrina/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Constricción , Femenino , Fibroblastos/patología , Fibroblastos/ultraestructura , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Cardiovasculares , Miocardio/patología , Comunicación Paracrina/genética , Ratas , Ratas Wistar , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Remodelación Ventricular , Proteína de Unión al GTP rhoA/metabolismo , Red trans-Golgi/metabolismo , Red trans-Golgi/ultraestructura
19.
Mol Med ; 21(1): 803-815, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26349059

RESUMEN

Erythropoietin (EPO) exerts potent neuroprotective, neuroregenerative and procognitive functions. However, unequivocal demonstration of erythropoietin receptor (EPOR) expression in brain cells has remained difficult since previously available anti-EPOR antibodies (EPOR-AB) were unspecific. We report here a new, highly specific, polyclonal rabbit EPOR-AB directed against different epitopes in the cytoplasmic tail of human and murine EPOR and its characterization by mass spectrometric analysis of immuno-precipitated endogenous EPOR, Western blotting, immunostaining and flow cytometry. Among others, we applied genetic strategies including overexpression, Lentivirus-mediated conditional knockout of EpoR and tagged proteins, both on cultured cells and tissue sections, as well as intracortical implantation of EPOR-transduced cells to verify specificity. We show examples of EPOR expression in neurons, oligodendroglia, astrocytes and microglia. Employing this new EPOR-AB with double-labeling strategies, we demonstrate membrane expression of EPOR as well as its localization in intracellular compartments such as the Golgi apparatus. Moreover, we show injury-induced expression of EPOR. In mice, a stereotactically applied stab wound to the motor cortex leads to distinct EpoR expression by reactive GFAP-expressing cells in the lesion vicinity. In a patient suffering from epilepsy, neurons and oligodendrocytes of the hippocampus strongly express EPOR. To conclude, this new analytical tool will allow neuroscientists to pinpoint EPOR expression in cells of the nervous system and to better understand its role in healthy conditions, including brain development, as well as under pathological circumstances, such as upregulation upon distress and injury.

20.
Stem Cells ; 32(9): 2480-91, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24806289

RESUMEN

The role of erythropoietin (Epo) in myocardial repair after infarction remains inconclusive. We observed high Epo receptor (EPOR) expression in cardiac progenitor cells (CPCs). Therefore, we aimed to characterize these cells and elucidate their contribution to myocardial regeneration on Epo stimulation. High EPOR expression was detected during murine embryonic heart development followed by a marked decrease until adulthood. EPOR-positive cells in the adult heart were identified in a CPC-enriched cell population and showed coexpression of stem, mesenchymal, endothelial, and cardiomyogenic cell markers. We focused on the population coexpressing early (TBX5, NKX2.5) and definitive (myosin heavy chain [MHC], cardiac Troponin T [cTNT]) cardiomyocyte markers. Epo increased their proliferation and thus were designated as Epo-responsive MHC expressing cells (EMCs). In vitro, EMCs proliferated and partially differentiated toward cardiomyocyte-like cells. Repetitive Epo administration in mice with myocardial infarction (cumulative dose 4 IU/g) resulted in an increase in cardiac EMCs and cTNT-positive cells in the infarcted area. This was further accompanied by a significant preservation of cardiac function when compared with control mice. Our study characterized an EPO-responsive MHC-expressing cell population in the adult heart. Repetitive, moderate-dose Epo treatment enhanced the proliferation of EMCs resulting in preservation of post-ischemic cardiac function.


Asunto(s)
Eritropoyetina/farmacología , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Receptores de Eritropoyetina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos C57BL , Ratas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA